US8703795B2 - Thienopyridine compounds, and methods of use thereof - Google Patents

Thienopyridine compounds, and methods of use thereof Download PDF

Info

Publication number
US8703795B2
US8703795B2 US11/367,969 US36796906A US8703795B2 US 8703795 B2 US8703795 B2 US 8703795B2 US 36796906 A US36796906 A US 36796906A US 8703795 B2 US8703795 B2 US 8703795B2
Authority
US
United States
Prior art keywords
thieno
pyridine
hydroxy
substituted
amino
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active, expires
Application number
US11/367,969
Other languages
English (en)
Other versions
US20060199836A1 (en
Inventor
Eric D. Turtle
Lee A. Flippin
Michael P. Arend
Heng Cheng
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Fibrogen Inc
Original Assignee
Fibrogen Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Fibrogen Inc filed Critical Fibrogen Inc
Priority to US11/367,969 priority Critical patent/US8703795B2/en
Publication of US20060199836A1 publication Critical patent/US20060199836A1/en
Assigned to FIBROGEN, INC. reassignment FIBROGEN, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CHENG, HENG, TURTLE, ERIC D., AREND, MICHAEL P., FLIPPIN, LEE A.
Priority to US14/249,281 priority patent/US9000006B2/en
Application granted granted Critical
Publication of US8703795B2 publication Critical patent/US8703795B2/en
Assigned to WILMINGTON TRUST, NATIONAL ASSOCIATION reassignment WILMINGTON TRUST, NATIONAL ASSOCIATION SECURITY INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: FIBROGEN, INC.
Active legal-status Critical Current
Adjusted expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D495/00Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms
    • C07D495/02Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D495/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/06Anti-spasmodics, e.g. drugs for colics, esophagic dyskinesia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/06Antianaemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/12Antihypertensives
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains two hetero rings

Definitions

  • This invention relates to novel compounds capable of modulating the stability and/or activity of hypoxia inducible factor (HIF).
  • HIF hypoxia inducible factor
  • Hypoxia inducible factor is a basic helix-loop-helix (bHLH) PAS (Per/Arnt/Sim) transcriptional activator that mediates changes in gene expression in response to changes in cellular oxygen concentration.
  • HIF is a heterodimer containing an oxygen-regulated ⁇ -subunit (HIF ⁇ ), and a constitutively expressed ⁇ -subunit (HIF ⁇ ), also known as aryl hydrocarbon receptor nuclear transporter (ARNT).
  • oxygenated (normoxic) cells HIF ⁇ subunits are rapidly degraded by a mechanism that involves ubiquitination by the von Hippel-Lindau tumor suppressor (pVHL) E3 ligase complex.
  • pVHL von Hippel-Lindau tumor suppressor
  • HIF ⁇ is not degraded, and an active HIF ⁇ / ⁇ complex accumulates in the nucleus, and activates the expression of several genes including glycolytic enzymes, glucose transporters, erythropoietin (EPO), and vascular endothelial growth factor (VEGF).
  • EPO erythropoietin
  • VEGF vascular endothelial growth factor
  • HIF ⁇ HIF ⁇ levels are elevated in most cells in response to hypoxia, and HIF ⁇ is induced in vivo when animals are subjected to anemia or hypoxia. HIF ⁇ levels rise within a few hours after the onset of hypoxia, and induce numerous beneficial cellular processes including cytoprotective effects, enhanced erythropoiesis, and physiological adaptation to ischemic or hypoxic states. Induction of HIF ⁇ is potentially beneficial in conditions such as myocardial acute ischemia, and early infarction, pulmonary hypertension, inflammation, and anemia.
  • HIF ⁇ levels are also increased by a number of factors that mimic hypoxia, including iron chelators such as desferrioxamine (DFO), and divalent metal salts such as CoCl 2 .
  • iron chelators such as desferrioxamine (DFO)
  • DFO desferrioxamine
  • CoCl 2 divalent metal salts
  • compounds originally identified as inhibitors of procollagen prolyl hydroxylase enzymes have been found to stabilize HIF ⁇ . Examples of such compounds can be found, e.g., in Majamaa et al. (1984) Eur J Biochem 138:239-245; Majamaa et al. (1985) Biochem J 229:127-133; Kivirikko, and Myllyharju (1998) Matrix Biol 16:357-368; Bickel et al. (1998) Hepatology 28:404-411; Friedman et al.
  • HIF hypoxia
  • This invention is directed to novel compounds, and methods that can modulate the stability and/or activity of hypoxia inducible factor (HIF).
  • HIF hypoxia inducible factor
  • q is 0 or 1
  • one of X or Y is —S—, and the other is ⁇ C(R 7 )—;
  • R 1 is selected from the group consisting of hydroxy, alkoxy, substituted alkoxy, acyloxy, cycloalkoxy, substituted cycloalkoxy, aryloxy, substituted aryloxy, heteroaryloxy, substituted heteroaryloxy, heterocyclyloxy, substituted heterocyclyloxy, mercapto, thioether, amino, substituted amino, and aminoacyl;
  • R 2 is selected from the group consisting of hydrogen, deuterium, and methyl
  • R 3 is selected from the group consisting of hydrogen, deuterium, alkyl, and substituted alkyl
  • R 4 is selected from the group consisting of hydrogen, alkyl, and substituted alkyl
  • R 5 is selected from the group consisting of hydrogen, halo, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, cycloalkoxy, substituted cycloalkoxy, amino, substituted amino, aminoacyl, aryl, substituted aryl, aryloxy, substituted aryloxy, heteroaryl, substituted heteroaryl, heteroaryloxy, substituted heteroaryloxy, heterocyclyl, substituted heterocyclyl, heterocyclyloxy, substituted heterocyclyloxy, thioether, cyano, and acyl;
  • R 6 and R 7 are independently selected from the group consisting of hydrogen, hydroxy, cyano, halo, nitro, acyl, amino, substituted amino, acylamino, sulfonyl, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, alkoxy, substituted alkoxy, aryl, substituted aryl, aryloxy, substituted aryloxy, thioether, arylthio, substituted arylthio, heteroaryl, and substituted heteroaryl;
  • the invention also provides pharmaceutical compositions comprising one or more compounds of Formula I and a pharmaceutically acceptable excipient.
  • the composition further comprises at least one additional therapeutic agent.
  • the agent is selected from the group consisting of vitamin B12, folic acid, ferrous sulfate, recombinant human erythropoietin and an erythropoiesis stimulating protein (ESP).
  • ESP erythropoiesis stimulating protein
  • the invention is also directed to methods of treating, pretreatng, or delaying onset of a condition mediated at least in part by hypoxia inducible factor (HIF) and/or erythropoietin (EPO), comprising administering to a patient, a therapeutically effective amount of a compound of Formula I.
  • HIF hypoxia inducible factor
  • EPO erythropoietin
  • q is 0 or 1
  • one of X or Y is —S—, and the other is ⁇ C(R 7 )—;
  • R 1 is selected from the group consisting of hydroxy, alkoxy, substituted alkoxy, acyloxy, cycloalkoxy, substituted cycloalkoxy, aryloxy, substituted aryloxy, heteroaryloxy, substituted heteroaryloxy, heterocyclyloxy, substituted heterocyclyloxy, mercapto, thioether, amino, substituted amino, and aminoacyl;
  • R 2 is selected from the group consisting of hydrogen, deuterium, and methyl
  • R 3 is selected from the group consisting of hydrogen, deuterium, alkyl, and substituted alkyl
  • R 4 is selected from the group consisting of hydrogen, alkyl, and substituted alkyl
  • R 5 is selected from the group consisting of hydrogen, halo, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, alkoxy, substituted alkoxy, cycloalkyl, substituted cycloalkyl, cycloalkoxy, substituted cycloalkoxy, amino, substituted amino, aminoacyl, aryl, substituted aryl, aryloxy, substituted aryloxy, heteroaryl, substituted heteroaryl, heteroaryloxy, substituted heteroaryloxy, heterocyclyl, substituted heterocyclyl, heterocyclyloxy, substituted heterocyclyloxy, thioether, cyano, and acyl;
  • R 6 and R 7 are independently selected from the group consisting of hydrogen, hydroxy, cyano, halo, nitro, acyl, amino, substituted amino, acylamino, sulfonyl, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, alkoxy, substituted alkoxy, aryl, substituted aryl, aryloxy, substituted aryloxy, thioether, arylthio, substituted arylthio, heteroaryl, and substituted heteroaryl;
  • this invention relates to compounds of formula Ia:
  • R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , and q are as defined above.
  • the invention relates to compounds of formula Ib:
  • R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , and q are as defined above.
  • q is 0.
  • R 1 is hydroxy. In particular embodiments wherein R 1 is hydroxy, R 2 , R 3 , and R 4 are all hydrogen.
  • R 5 is selected from the group consisting of hydrogen, alkyl, halo, aryl, substituted aryl, cyano, alkynyl, and heteroaryl. In particular embodiments, R 5 is selected from hydrogen, methyl, bromo, chloro, phenyl, fluorophenyl, cyano, ethynyl, furanyl, and thienyl.
  • R 6 is selected from the group consisting of hydrogen, halo, alkyl, substituted alkyl, alkenyl, substituted alkenyl, aryl, substituted aryl, aryloxy, arylthio, and cyano.
  • R 6 is selected from the group consisting of hydrogen, bromo, methyl, phenyl, trifluoromethylphenyl, phenoxyphenyl, fluorophenyl, phenylsulfanyl, phenoxy, phenethyl, phenylethenyl, and cyano.
  • R 7 is hydrogen, aryl, or substituted aryl. In particular embodiments, R 7 is hydrogen, phenyl, or fluorophenyl.
  • the present invention relates to compounds of formula Ia wherein
  • the present invention relates to compounds of formula Ia wherein
  • R 1 is hydroxy
  • R 2 , R 3 , and R 4 are hydrogen
  • R 5 is halo, alkyl, alkynyl, cyano, aryl, substituted aryl, or heteroaryl;
  • R 6 is hydrogen, halo, alkyl, aryl, substituted aryl or cyano
  • R 7 is hydrogen, aryl, or substituted aryl
  • the invention relates to compounds of formula Ib wherein
  • R 1 is hydroxy
  • R 2 , R 3 , and R 4 are hydrogen
  • R 5 is hydrogen, halo, alkyl, or aryl
  • R 6 is halo, alkyl, aryl, substituted aryl, or arylthio
  • R 7 is hydrogen or substituted aryl
  • the invention relates to compounds of formula Ib wherein
  • R 1 is hydroxy
  • R 2 , R 3 , and R 4 are hydrogen
  • R 5 is hydrogen, halo, alkyl, aryl, substituted aryl, heteroaryl, alkynyl, or cyano;
  • R 6 is hydrogen, halo, alkyl, substituted aryl, arylsulfanyl, or cyano;
  • R 7 is hydrogen or substituted aryl
  • Compounds included within the scope of this invention include, for example, [(2-bromo-4-hydroxy-thieno[2,3-c]pyridine-5-carbonyl)-amino]-acetic acid; [(2-bromo-7-hydroxy-thieno[3,2-c]pyridine-6-carbonyl)-amino]-acetic acid; ⁇ [4-hydroxy-2-(4-methoxy-phenyl)-thieno[2,3-c]pyridine-5-carbonyl]-amino ⁇ -acetic acid; ⁇ [7-hydroxy-2-(4-methoxy-phenyl)-thieno[3,2-c]pyridine-6-carbonyl]amino ⁇ -acetic acid; [(4-hydroxy-2,7-dimethyl-thieno[2,3-c]pyridine-5-carbonyl)-amino]-acetic acid; [(7-hydroxy-2,4-dimethyl-thieno[3,2-c]pyridine-6-carbonyl)-amino]
  • the invention also provides for use of a compound of formula I, Ia, and/or Ib for the manufacture of a medicament for use in treating various conditions or disorders as described below.
  • a pharmaceutical composition comprising a pharmaceutically acceptable excipient or carrier, and a therapeutically effective amount of at least one compound selected from the group consisting of formulae I, Ia, and/or Ib.
  • the invention also contemplates the medicament or composition further comprising at least one additional therapeutic agent selected from the group including, but not limited to, vitamin B12, ferrous sulfate, folic acid, and/or recombinant erythropoietin or an erythropoiesis stimulating protein (ESP).
  • at least one additional therapeutic agent selected from the group including, but not limited to, vitamin B12, ferrous sulfate, folic acid, and/or recombinant erythropoietin or an erythropoiesis stimulating protein (ESP).
  • ESP erythropoiesis stimulating protein
  • the invention is also directed to use of a compound, or composition or medicament thereof, to treat, pretreat, or delay onset of a condition mediated at least in part by hypoxia inducible factor (HIF) and/or erythropoietin (EPO).
  • the use comprises administering to a mammalian patient a therapeutically effective amount of a medicament or pharmaceutical composition comprising one or more compounds of formulae I, Ia, and/or Ib.
  • the condition can be selected from the group consisting of anemic disorders; neurological disorders and/or injuries including cases of stroke, trauma, epilepsy, and neurodegenerative disease; myocardial infarction, liver ischemia, renal ischemia, peripheral vascular disorders, ulcers, burns, and chronic wounds; pulmonary embolism; and ischemic-reperfusion injury.
  • the invention is also directed to a method of inhibiting the activity of at least one hydroxylase enzyme which modifies the alpha subunit of hypoxia inducible factor.
  • the method comprises contacting the enzyme with an inhibiting effective amount of one or more compounds selected from the group comprising compounds of formulae I, Ia, and/or Ib.
  • HIF ⁇ refers to the alpha subunit of hypoxia inducible factor protein.
  • HIF ⁇ may be any human or other mammalian protein, or fragment thereof, including, but not limited to, human HIF-1 ⁇ (Genbank Accession No. Q16665), HIF-2 ⁇ (Genbank Accession No. AAB41495), and HIF-3 ⁇ (Genbank Accession No. AAD22668); murine HIF-1 ⁇ (Genbank Accession No. Q61221), HIF-2 ⁇ (Genbank Accession No. BAA20130, and AAB41496), and HIF-3 ⁇ (Genbank Accession No. AAC72734); rat HIF-1 ⁇ (Genbank Accession No. CAA70701), HIF-2 ⁇ (Genbank Accession No.
  • HIF ⁇ may also be any non-mammalian protein or fragment thereof, including Xenopus laevis HIF-1 ⁇ (Genbank Accession No. CAB96628), Drosophila melanogaster HIF-1 ⁇ (Genbank Accession No. JC4851), and chicken HIF-1 ⁇ (Genbank Accession No. BAA34234).
  • a fragment of HIF ⁇ includes any fragment retaining at least one functional or structural characteristic of HIF ⁇ .
  • Fragments of HIF ⁇ include, e.g., the regions defined by human HIF-1 ⁇ from amino acids 401 to 603 (Huang et al., supra), amino acid 531 to 575 (Jiang et al. (1997) J. Biol. Chem 272:19253-19260), amino acid 556 to 575 (Tanimoto et al., supra), amino acid 557 to 571 (Srinivas et al. (1999) Biochem Biophys Res. Commun 260:557-561), and amino acid 556 to 575 (Ivan, and Kaelin (2001) Science 292:464-468).
  • HIF ⁇ fragments include any fragment containing at least one occurrence of the motif LXXLAP, e.g., as occurs in the human HIF-1 ⁇ native sequence from L 397 to P 402 , and from L 559 to P 564 .
  • HIF PH refers to any enzyme capable of hydroxylating a proline residue in the HIF protein.
  • the proline residue hydroxylated by HIF PH includes the proline found within the motif LXXLAP.
  • HIF PH includes members of the Egl-Nine (EGLN) gene family described by Taylor (2001, Gene 275:125-132), and characterized by Aravind, and Koonin (2001, Genome Biol 2: RESEARCH 0007), Epstein et al. (2001, Cell 107:43-54), and Bruick and McKnight (2001, Science 294:1337-1340).
  • HIF PH2 as used in assays described herein, may be selected from human EGLN1 (hEGLN1, GenBank Accession No. AAG33965; Dupuy et al. (2000) Genomics 69:348-54), mouse EGLN1 (GenBank Accession No. CAC42515), and rat EGLN1 (GenBank Accession No. P59722).
  • another HIF PH may be used in the assay.
  • HIF PH enzymes include, but are not limited to, human EGLN2 isoform 1 (GenBank Accession No. CAC42510; Taylor, supra), human EGLN2 isoform 3 (GenBank Accession No. NP — 542770), mouse EGLN2 (GenBank Accession No.
  • EGLN may include Caenorhabditis elegans EGL-9 (GenBank Accession No. AAD56365) and Drosophila melanogaster CG1114 gene product (GenBank Accession No. AAF52050).
  • HIF PH also includes any fragment of the foregoing full-length proteins that retain at least one structural or functional characteristic.
  • anemia refers to any abnormality in hemoglobin or erythrocytes that leads to reduced oxygen levels in the blood.
  • Anemia can be associated with abnormal production, processing, or performance of erythrocytes and/or hemoglobin.
  • anemia refers to any reduction in the number of red blood cells and/or level of hemoglobin in blood relative to normal blood levels.
  • Anemia can arise due to conditions such as acute or chronic kidney disease, infections, inflammation, cancer, irradiation, toxins, diabetes, and surgery. Infections may be due to, e.g., virus, bacteria, and/or parasites, etc. Inflammation may be due to infection or autoimmune disorders, such as rheumatoid arthritis, etc. Anemia can also be associated with blood loss due to, e.g., stomach ulcer, duodenal ulcer, hemorrhoids, cancer of the stomach or large intestine, trauma, injury, surgical procedures, etc. Anemia is further associated with radiation therapy, chemotherapy, and kidney dialysis.
  • Anemia is also associated with HIV-infected patients undergoing treatment with azidothymidine (zidovudine) or other reverse transcriptase inhibitors, and can develop in cancer patients undergoing chemotherapy, e.g., with cyclic cisplatin- or non-cisplatin-containing chemotherapeutics.
  • Aplastic anemia and myelodysplastic syndromes are diseases associated with bone marrow failure that result in decreased production of erythrocytes.
  • anemia can result from defective or abnormal hemoglobin or erythrocytes, such as in disorders including microcytic anemia, hypochromic anemia, etc.
  • Anemia can result from disorders in iron transport, processing, and utilization, see, e.g., sideroblastic anemia, etc.
  • anemic conditions and “anemic disorders” refer to any condition, disease, or disorder associated with anemia. Such disorders include, but are not limited to, those disorders listed above. Anemic disorders further include, but are not limited to, aplastic anemia, autoimmune hemolytic anemia, bone marrow transplantation, Churg-Strauss syndrome, Diamond Blackfan anemia, Fanconi's anemia, Felty syndrome, graft versus host disease, hematopoietic stem cell transplantation, hemolytic uremic syndrome, myelodysplastic syndrome, nocturnal paroxysmal hemoglobinuria, osteomyelofibrosis, pancytopenia, pure red-cell aplasia, purpura Schoenlein-Henoch, sideroblastic anemia, refractory anemia with excess of blasts, rheumatoid arthritis, Shwachman syndrome, sickle cell disease, thalassemia major, thalassemia minor, thrombocytopenic purpura, etc.
  • erythropoietin-associated conditions is used inclusively and refers to any condition associated with below normal, abnormal, or inappropriate modulation of erythropoietin.
  • Erythropoietin-associated conditions include any condition wherein an increase in EPO level would provide therapeutic benefit. Levels of erythropoietin associated with such conditions can be determined by any measure accepted and utilized by those of skill in the art. Erythropoietin-associated conditions include anemic conditions such as those described above.
  • Erythropoietin-associated conditions further include neurological disorders and/or injuries, including cases of stroke, trauma, epilepsy, neurodegenerative disease and the like, wherein erythropoietin may provide a neuroprotective effect.
  • Neurodegenerative diseases contemplated by the invention include Alzheimer's disease, Parkinson's disease, Huntington's disease, and the like.
  • erythropoietin refers to any recombinant or naturally occurring erythropoietin or ESP including, e.g., human erythropoietin (GenBank Accession No. AAA52400; Lin et al. (1985) Proc Nat'l Acad.
  • EPOETIN human recombinant erythropoietin Amgen, Inc., Thousand Oaks Calif.
  • ARANESP human recombinant erythropoietin Amgen
  • PROCRIT human recombinant erythropoietin Ortho Biotech Products, L.P., Raritan N.J.
  • glycosylated erythropoietin such as those described in U.S. Pat. No. 6,930,086 (which is incorporated by reference), etc.
  • alkyl refers to saturated monovalent hydrocarbyl groups having from 1 to 10 carbon atoms, preferably from 1 to 5 carbon atoms, and more preferably 1 to 3 carbon atoms. This term is exemplified by groups such as methyl, ethyl, n-propyl, iso-propyl, n-butyl, t-butyl, n-pentyl, and the like.
  • substituted alkyl refers to an alkyl group, of from 1 to 10 carbon atoms, preferably, 1 to 5 carbon atoms, having from 1 to 5 substituents, preferably 1 to 3 substituents, independently selected from the group consisting of alkoxy, substituted alkoxy, acyl, acylamino, acyloxy, amino, substituted amino, aminoacyl, aminocarbonylamino, aminothiocarbonylamino, aminocarbonyloxy, aryl, substituted aryl, aryloxy, substituted aryloxy, aryloxyaryl, substituted aryloxyaryl, cyano, halogen, hydroxyl, nitro, oxo, thioxo, carboxyl, carboxyl esters, cycloalkyl, substituted cycloalkyl, thiol, alkylthio, substituted alkylthio, arylthio, substituted arylthio, cycloalkylthi
  • alkoxy refers to the group “alkyl-O-” which includes, by way of example, methoxy, ethoxy, n-propoxy, iso-propoxy, n-butoxy, t-butoxy, sec-butoxy, n-pentoxy, and the like.
  • substituted alkoxy refers to the group “substituted alkyl-O-”.
  • acyl refers to the groups H—C(O)—, alkyl-C(O)—, substituted alkyl-C(O)—, alkenyl-C(O)—, substituted alkenyl-C(O)—, alkynyl-C(O)—, substituted alkynyl-C(O)—, cycloalkyl-C(O)—, substituted cycloalkyl-C(O)—, aryl-C(O)—, substituted aryl-C(O)—, heteroaryl-C(O)—, substituted heteroaryl-C(O), heterocyclic-C(O)—, and substituted heterocyclic-C(O)— provided that a nitrogen atom of the heterocyclic or substituted heterocyclic is not bound to the —C(O)— group wherein alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cyclo
  • aminoacyl and the prefix “carbamoyl” or “carboxamide” or “substituted carbamoyl” or “substituted carboxamide” refers to the group —C(O)NR 42 R 42 where each R 42 is independently selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, substituted aryl, cycloalkyl, substituted cycloalkyl, heteroaryl, substituted heteroaryl, heterocyclic, and substituted heterocyclic; or where each R 42 is joined to form together with the nitrogen atom a heterocyclic or substituted heterocyclic wherein alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, aryl, substituted aryl, hetero
  • acyloxy refers to the groups alkyl-C(O)O—, substituted alkyl-C(O)O—, alkenyl-C(O)O—, substituted alkenyl-C(O)O—, alkynyl-C(O)O—, substituted alkynyl-C(O)O—, aryl-C(O)O—, substituted aryl-C(O)O—, cycloalkyl-C(O)O—, substituted cycloalkyl-C(O)O—, heteroaryl-C(O)O—, substituted heteroaryl-C(O)O—, heterocyclic-C(O)O—, and substituted heterocyclic-C(O)O—, wherein alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, aryl, substituted aryl,
  • alkenyl refers to a vinyl unsaturated monovalent hydrocarbyl group having from 2 to 6 carbon atoms, and preferably 2 to 4 carbon atoms, and having at least 1, and preferably from 1 to 2 sites of vinyl (>C ⁇ C ⁇ ) unsaturation.
  • groups are exemplified by vinyl (ethen-1-yl), allyl, but-3-enyl and the like.
  • substituted alkenyl refers to alkenyl groups having from 1 to 3 substituents, and preferably 1 to 2 substituents, selected from the group consisting of alkoxy, substituted alkoxy, acyl, acylamino, acyloxy, amino, substituted amino, aminoacyl, aryl, substituted aryl, aryloxy, substituted aryloxy, cyano, halogen, hydroxyl, nitro, carboxyl, carboxyl esters, cycloalkyl, substituted cycloalkyl, heteroaryl, substituted heteroaryl, heterocyclic, and substituted heterocyclic.
  • This term includes both E (cis) and Z (trans) isomers as appropriate. It also includes mixtures of both E and Z components.
  • alkynyl refers to an acetylenic unsaturated monovalent hydrocarbyl groups having from 2 to 6 carbon atoms, and preferably 2 to 3 carbon atoms, and having at least 1, and preferably from 1 to 2 sites of acetylenic (—C ⁇ C—) unsaturation. This group is exemplified by ethyn-1-yl, propyn-1-yl, propyn-2-yl, and the like.
  • substituted alkynyl refers to alkynyl groups having from 1 to 3 substituents, and preferably 1 to 2 substituents, selected from the group consisting of alkoxy, substituted alkoxy, acyl, acylamino, acyloxy, amino, substituted amino, aminoacyl, aryl, substituted aryl, aryloxy, substituted aryloxy, cyano, halogen, hydroxyl, nitro, carboxyl, carboxyl esters, cycloalkyl, substituted cycloalkyl, heteroaryl, substituted heteroaryl, heterocyclic, and substituted heterocyclic.
  • amino refers to the group —NH 2 .
  • substituted amino refers to the group —NR 41 R 41 , where each R 41 is independently selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic, substituted heterocyclic, —SO 2 -alkyl, —SO 2 -substituted alkyl, —SO 2 -alkenyl, —SO 2 -substituted alkenyl, —SO 2 -cycloalkyl, —SO 2 -substituted cycloalkyl, —SO 2 -aryl, —SO 2 -substituted aryl, —SO 2 -heteroaryl, —SO 2 -substituted heteroaryl, —
  • acylamino refers to the groups —NR 45 C(O)alkyl, —NR 45 C(O)substituted alkyl, —NR 45 C(O)cycloalkyl, —NR 45 C(O)substituted cycloalkyl, —NR 45 C(O)alkenyl, —NR 45 C(O)substituted alkenyl, —NR 45 C(O)alkynyl, —NR 45 C(O)substituted alkynyl, —NR 45 C(O)aryl, —NR 45 C(O)substituted aryl, —NR 45 C(O)heteroaryl, —NR 45 C(O)substituted heteroaryl, —NR 45 C(O)heterocyclic, and —NR 45 C(O)substituted heterocyclic where R 45 is hydrogen or alkyl, and wherein alkyl, substituted alkyl, alken
  • oxycarbonylamino refers to the groups —NR 46 C(O)O-alkyl, —NR 46 C(O)O-substituted alkyl, —NR 46 C(O)O-alkenyl, —NR 46 C(O)O-substituted alkenyl, —NR 46 C(O)O-alkynyl, —NR 46 C(O)O-substituted alkynyl, —NR 46 C(O)O-cycloalkyl, —NR 46 C(O)O-substituted cycloalkyl, —NR 46 C(O)O-aryl, —NR 46 C(O)O-substituted aryl, —NR 46 C(O)O-heteroaryl, —NR 46 C(O)O-substituted heteroaryl, —NR 46 C(O)O-heterocyclic, and —NR 46 C(O)O-substituted
  • oxythiocarbonylamino refers to the groups —NR 46 C(S)O-alkyl, —NR 46 C(S)O-substituted alkyl, —NR 46 C(S)O-alkenyl, —NR 46 C(S)O-substituted alkenyl, —NR 46 C(S)O-alkynyl, —NR 46 C(S)O-substituted alkynyl, —NR 46 C(S)O-cycloalkyl, —NR 46 C(S)O-substituted cycloalkyl, —NR 46 C(S)O-aryl, —NR 46 C(S)O-substituted aryl, —NR 46 C(S)O-heteroaryl, —NR 46 C(S)O-substituted heteroaryl, —NR 46 C(S)O-heterocyclic, and —NR 46 C(S)O-sub-sub
  • aminocarbonyloxy or as a prefix “carbamoyloxy” or “substituted carbamoyloxy” refers to the groups —OC(O)NR 47 R 47 where each R 47 is independently selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocyclic, and substituted heterocyclic; or where each R 47 is joined to form, together with the nitrogen atom, a heterocyclic or substituted heterocyclic, and wherein alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, aryl, substituted aryl, heteroaryl, substituted heteroary
  • aminocarbonylamino refers to the group —NR 49 C(O)NR 49 — where each R 49 is independently selected from the group consisting of hydrogen and alkyl.
  • aminothiocarbonylamino refers to the group —NR 49 C(S)NR 49 — where each R 49 is independently selected from the group consisting of hydrogen and alkyl.
  • aryl refers to a monovalent aromatic carbocyclic group of from 6 to 14 carbon atoms having a single ring (e.g., phenyl) or multiple condensed rings (e.g., naphthyl or anthryl) which condensed rings may or may not be aromatic (e.g., 2-benzoxazolinone, 2H-1,4-benzoxazin-3(4H)-one-7-yl, and the like) provided that the point of attachment is the aryl group.
  • Preferred aryls include phenyl, and naphthyl.
  • substituted aryl refers to aryl groups, as defined herein, which are substituted with from 1 to 4, preferably 1 to 3, substituents selected from the group consisting of hydroxy, acyl, acylamino, acyloxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, amidino, amino, substituted amino, aminoacyl, aminocarbonyloxy, aminocarbonylamino, aminothiocarbonylamino, aryl, substituted aryl, aryloxy, substituted aryloxy, cycloalkoxy, substituted cycloalkoxy, heteroaryloxy, substituted heteroaryloxy, heterocyclyloxy, substituted heterocyclyloxy, carboxyl, carboxyl esters, cyano, thiol, alkylthio, substituted alkylthio, arylthio, substituted substituted alkyloxy,
  • aryloxy refers to the group aryl-O— that includes, by way of example, phenoxy, naphthoxy, and the like.
  • substituted aryloxy refers to substituted aryl-O— groups.
  • aryloxyaryl refers to the group -aryl-O-aryl.
  • substituted aryloxyaryl refers to aryloxyaryl groups substituted with from 1 to 3 substituents on either or both aryl rings as defined above for substituted aryl.
  • carboxyl refers to —COOH or salts thereof.
  • carboxyl esters refers to the groups —C(O)O-alkyl, —C(O)O-substituted alkyl, —C(O)O-alkenyl, —C(O)O-substituted alkenyl, —C(O)O-alkynyl, —C(O)O-substituted alkynyl, —C(O)O-aryl, —C(O)O-substituted aryl, —C(O)O-heteroaryl, —C(O)O-substituted heteroaryl, —C(O)O-heterocyclic, and —C(O)O-substituted heterocyclic.
  • cycloalkyl refers to cyclic alkyl groups of from 3 to 10 carbon atoms having single or multiple cyclic rings including, by way of example, adamantyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclooctyl, and the like.
  • substituted cycloalkyl refers to a cycloalkyl group, having from 1 to 5 substituents selected from the group consisting of oxo ( ⁇ O), thioxo ( ⁇ S), alkyl, substituted alkyl, alkoxy, substituted alkoxy, acyl, acylamino, acyloxy, amino, substituted amino, aminoacyl, aryl, substituted aryl, aryloxy, substituted aryloxy, cyano, halogen, hydroxy, nitro, carboxyl, carboxyl esters, cycloalkyl, substituted cycloalkyl, heteroaryl, substituted heteroaryl, heterocyclic, and substituted heterocyclic.
  • substituents selected from the group consisting of oxo ( ⁇ O), thioxo ( ⁇ S), alkyl, substituted alkyl, alkoxy, substituted alkoxy, acyl, acylamino, acyloxy, amino, substituted amino, aminoacyl
  • cycloalkoxy refers to —O-cycloalkyl groups.
  • substituted cycloalkoxy refers to —O-substituted cycloalkyl groups.
  • halo or halogen refers to fluoro, chloro, bromo, and iodo, and preferably is fluoro or chloro.
  • heteroaryl refers to an aromatic group of from 1 to 15 carbon atoms, preferably from 1 to 10 carbon atoms, and 1 to 4 heteroatoms selected from the group consisting of oxygen, nitrogen, and sulfur within the ring.
  • Such heteroaryl groups can have a single ring (e.g., pyridinyl, furyl, or thienyl) or multiple condensed rings (e.g., indolizinyl or benzothienyl).
  • the nitrogen and/or sulfur ring atoms can optionally be oxidized to provide for the N-oxide or the sulfoxide, and sulfone derivatives.
  • Preferred heteroaryls include pyridinyl, pyrrolyl, indolyl, thiophenyl, thienyl, and furyl.
  • substituted heteroaryl refers to heteroaryl groups that are substituted with from 1 to 3 substituents selected from the same group of substituents defined for substituted aryl.
  • heteroaryloxy refers to the group —O-heteroaryl
  • substituted heteroaryloxy refers to the group —O-substituted heteroaryl
  • heterocycle refers to a saturated or unsaturated (but not aromatic) group having a single ring or multiple condensed rings, from 1 to 10 carbon atoms, and from 1 to 4 hetero atoms selected from the group consisting of nitrogen, sulfur or oxygen within the ring wherein, in fused ring systems, one or more of the rings can be aryl or heteroaryl provided that the point of attachment is at the heterocycle.
  • the nitrogen and/or sulfur ring atoms can optionally be oxidized to provide for the N-oxide or the sulfoxide, and sulfone derivatives.
  • substituted heterocyclic refers to heterocycle groups that are substituted with from 1 to 3 of the same substituents as defined for substituted cycloalkyl.
  • heterocycles and heteroaryls include, but are not limited to, azetidine, pyrrole, imidazole, pyrazole, pyridine, pyrazine, pyrimidine, pyridazine, indolizine, isoindole, indole, dihydroindole, indazole, purine, quinolizine, isoquinoline, quinoline, phthalazine, naphthylpyridine, quinoxaline, quinazoline, cinnoline, pteridine, carbazole, carboline, phenanthridine, acridine, phenanthroline, isothiazole, phenazine, isoxazole, phenoxazine, phenothiazine, imidazolidine, imidazoline, piperidine, piperazine, indoline, phthalimide, 1,2,3,4-tetrahydroisoquinoline, 4,5,6,
  • Heterocyclyloxy refers to the group —O-heterocyclic
  • substituted heterocyclyloxy refers to the group —O-substituted heterocyclic
  • Thiol or “mercapto” refers to the group —SH.
  • sulfonyl refers to the group —SO 2 H.
  • Alkylsulfanyl refers to the groups —S-alkyl where alkyl is as defined above.
  • Substituted alkylthio and “substituted alkylsulfanyl” refer to the group —S-substituted alkyl where alkyl is as defined above.
  • Cycloalkylthio or “cycloalkylsulfanyl” refers to the groups —S-cycloalkyl where cycloalkyl is as defined above.
  • Substituted cycloalkylthio refers to the group —S-substituted cycloalkyl where substituted cycloalkyl is as defined above.
  • Arylthio or “arylsulfanyl” refers to the group —S-aryl
  • substituted arylthio refers to the group —S-substituted aryl where aryl and substituted aryl are as defined above.
  • Heteroarylthio or “heteroarylsulfanyl” refers to the group —S-heteroaryl
  • substituted heteroarylthio refers to the group —S-substituted heteroaryl where heteroaryl and substituted heteroaryl are as defined above.
  • Heterocyclicthio refers to the group —S-heterocyclic
  • substituted heterocyclicthio refers to the group —S-substituted heterocyclic where heterocyclic, and substituted heterocyclic are as defined above.
  • amino acid refers to any of the naturally occurring amino acids, as well as synthetic analogs (e.g., D-stereoisomers of the naturally occurring amino acids, such as D-threonine), and derivatives thereof.
  • ⁇ -Amino acids comprise a carbon atom to which is bonded an amino group, a carboxyl group, a hydrogen atom, and a distinctive group referred to as a “side chain.”
  • the side chains of naturally occurring amino acids are well known in the art, and include, for example, hydrogen (e.g., as in glycine), alkyl (e.g., as in alanine, valine, leucine, isoleucine, proline), substituted alkyl (e.g., as in threonine, serine, methionine, cysteine, aspartic acid, asparagine, glutamic acid, glutamine, arginine, and lysine), arylalkyl (e.g., as in phenyla
  • Unnatural amino acids are also known in the art, as set forth in, for example, Williams, ed. (1989) Synthesis of Optically Active ⁇ -Amino Acids, Pergamon Press; Evans et al. (1990) J. Amer. Chem. Soc. 112:4011-4030; Pu et al. (1991) J. Amer. Chem. Soc. 56:1280-1283; Williams et al. (1991) J. Amer. Chem. Soc. 113:9276-9286; and all references cited therein.
  • the present invention includes the side chains of unnatural amino acids as well.
  • “Pharmaceutically acceptable salt” refers to pharmaceutically acceptable salts of a compound, which salts are derived from a variety of organic, and inorganic counter ions well known in the art, and include, by way of example only, sodium, potassium, calcium, magnesium, ammonium, tetraalkylammonium, and the like; and when the molecule contains a basic functionality, salts of organic or inorganic acids, such as hydrochloride, hydrobromide, tartrate, mesylate, acetate, maleate, oxalate, and the like.
  • prodrug refers to compounds of formula I, Ia, and Ib that include chemical groups which, in vivo, can be converted into the carboxylate group on the glycine or alanine substituent of the compounds and/or can be split off from the amide N-atom and/or can be split off from the 4-O atom of the thieno[2,3-c]pyridine or the 7-O atom of the thieno[3,2-c]pyridine; and/or can be split off from the N-atom of the pyridyl ring to provide for the active drug, a pharmaceutically acceptable salt thereof, or a biologically active metabolite thereof.
  • Suitable groups are well known in the art and particularly include: for the carboxylic acid moiety on the glycine or alanine substituent, a prodrug selected from, e.g., esters including, but not limited to, those derived from alkyl alcohols, substituted alkyl alcohols, hydroxy substituted aryls and heteroaryls and the like; amides, particularly amides derived from amines of the formula HNR 20 R 21 where R 20 and R 21 are independently hydrogen, alkyl, substituted alkyl, aryl, substituted aryl, and the like; hydroxymethyl, aldehyde and derivatives thereof; and for the pyridyl N atom, a prodrug selected from, e.g., N-oxides and N-alkyl derivatives.
  • esters including, but not limited to, those derived from alkyl alcohols, substituted alkyl alcohols, hydroxy substituted aryls and heteroaryls and the like
  • amides
  • excipient means an inert or inactive substance used in the production of pharmaceutical products or other tablets, including without limitation any substance used as a binder, disintegrant, coating, compression/encapsulation aid, cream or lotion, lubricant, parenteral, sweetener or flavoring, suspending/gelling agent, or wet granulation agent.
  • Binders include, e.g., carbopol, povidone, xanthan gum, etc.; coatings include, e.g., cellulose acetate phthalate, ethylcellulose, gellan gum, maltodextrin, etc.; compression/encapsulation aids include, e.g., calcium carbonate, dextrose, fructose dc, honey dc, lactose (anhydrate or monohydrate; optionally in combination with aspartame, cellulose, or microcrystalline cellulose), starch dc, sucrose, etc.; disintegrants include, e.g., croscarmellose sodium, gellan gum, sodium starch glycolate, etc.; creams and lotions include, e.g., maltodextrin, carrageenans, etc.; lubricants include, e.g., magnesium stearate, stearic acid, sodium stearyl fumarate, etc.; materials for chewable tablets include, e.g
  • impermissible substitution patterns e.g., methyl substituted with 5 fluoro groups or a hydroxyl group alpha to ethenylic or acetylenic unsaturation.
  • impermissible substitution patterns are well known to the skilled artisan.
  • the compounds of this invention can be prepared from readily available starting materials using, for example, the following general methods, and procedures. It will be appreciated that where typical or preferred process conditions (i.e., reaction temperatures, times, mole ratios of reactants, solvents, pressures, etc.) are given, other process conditions can also be used unless otherwise stated. Optimum reaction conditions may vary with the particular reactants or solvent used, but such conditions can be determined by one skilled in the art by routine optimization procedures.
  • protecting groups may be necessary to prevent certain functional groups from undergoing undesired reactions.
  • Suitable protecting groups for various functional groups as well as suitable conditions for protecting and deprotecting particular functional groups are well known in the art. For example, numerous protecting groups are described in T. W. Greene and G. M. Wuts (1999) Protecting Groups in Organic Synthesis, 3rd Edition, Wiley, New York, and references cited therein.
  • the compounds of this invention may contain one or more chiral centers. Accordingly, if desired, such compounds can be prepared or isolated as pure stereoisomers, i.e., as individual enantiomers or diastereomers, or as stereoisomer-enriched mixtures. All such stereoisomers (and enriched mixtures) are included within the scope of this invention, unless otherwise indicated. Pure stereoisomers (or enriched mixtures) may be prepared using, for example, optically active starting materials or stereoselective reagents well-known in the art. Alternatively, racemic mixtures of such compounds can be separated using, for example, chiral column chromatography, chiral resolving agents, and the like.
  • the starting materials for the following reactions are generally known compounds or can be prepared by known procedures or obvious modifications thereof.
  • many of the starting materials are available from commercial suppliers such as Aldrich Chemical Co. (Milwaukee, Wis., USA), Bachem (Torrance, Calif., USA), Emka-Chemce or Sigma (St. Louis, Mo., USA).
  • Scheme 1 illustrates a general method for the preparation of intermediates and compounds of this invention. These compounds are prepared from starting materials either known in the art or commercially available.
  • the carboxaldehyde substituent of compounds of the formula 102 is oxidized under conventional conditions, for example, by treatment with sodium chlorite, Jones reagent, or silver oxide, to provide for functionalized-3-methylthiophene-2-carboxylic acid, compounds of the formula 104.
  • R 6 or R 7 may be H, Br, Cl, I, NO 2 , F, aryl, and substituted aryl.
  • Subsequent oxidation of the 3-methyl substituent to the corresponding 3-carboxylic acid group of dicarboxylic acid compounds of the formula 106 proceed, again, by conventional methods using, for example, an excess of potassium permanganate as the oxidizing agent.
  • compounds of the formula 102 may be directly oxidized to compounds of the formula 106, for example, by treatment with excess potassium permanganate.
  • Esterification of the dicarboxylic acid groups of compounds of the formula 106 proceeds conventionally to provide for the corresponding diester of compounds of the formula 108.
  • esterification is accomplished by treatment of the di-acid with an activating species, such as thionyl chloride or oxalyl chloride, followed by addition of alcohol, such as methanol, ethanol, or butanol, where methanol is preferred, or for example, by treatment of 106 with a strong acid in the presence of excess alcohol.
  • an activating species such as thionyl chloride or oxalyl chloride
  • alcohol such as methanol, ethanol, or butanol, where methanol is preferred, or for example, by treatment of 106 with a strong acid in the presence of excess alcohol.
  • Mono-deesterification of compounds of the formula 108 proceeds using an equivalent of sodium hydroxide which provides for a mixture of thiophene-2,3-dicarboxylic acid 2-methyl ester and thiophene-2,3-dicarboxylic acid 3-methyl ester, compounds of the formula 110 and 112.
  • the free acid group of compounds of the formula 110 and 112 are amidated via conventional methods to provide for compounds of the formula 114 and 116.
  • Cyclization of compounds of the formula 114 and 116 in the presence of a suitable base, such as sodium alkoxide, where sodium n-butoxide in n-butanol is preferred, provides for functionalized-4,7-dihydroxythieno[2,3-c]pyridine-5-carboxylic acid alkyl ester and functionalized-4,7-dihydroxythieno[3,2-c]pyridine-6-carboxylic acid alkyl ester, compounds of the formula 118 and 120, where the n-butyl ester is preferred.
  • the hydroxyl group alpha to the nitrogen atom in the pyridinyl group of compounds of the formula 118 and 120 is substantially more reactive than the opposing hydroxyl group and, accordingly, allows for selective differentiation over the opposing hydroxyl group. Differentiation can be achieved by treating compounds of the formula 118 and 120 with an excess of a phosphorus oxyhalide, such as POCl 3 or POBr 3 , which results in halogenation to produce compounds of the formula 122 and 124 where R 5 is halogen.
  • Conventional amidation conditions using a compound of the formula H 2 NCR 2 R 3 C(O)OH or an ester thereof then provides for compounds of the formula 126 and 128 (after de-esterification if the ester is used).
  • Scheme 2 illustrates an alternative method for preparing intermediates useful in preparing compounds of this invention.
  • compounds of the formula 106 are converted first to the corresponding anhydride, compounds of the formula 130, by treatment with an excess of acetic anhydride.
  • the anhydride is converted to the corresponding N-substituted imide, compounds of the formula 132, by reaction with a glycine ester (GlyOR, where R may be, but is not limited to, methyl, ethyl, or n-butyl) followed by treatment with a carboxylic acid activating species, such as thionyl chloride, pivaloyl chloride, chloroformate derivatives, carbodiimides, or oxalyl chloride.
  • GlyOR glycine ester
  • Scheme 3 illustrates a second general route for the preparation of intermediates to compounds pertaining to this invention.
  • R 6 and R 7 may be hydrogen or halogen.
  • Compounds of the formula 134a and 134b where R may be, but is not limited to, methyl, ethyl, or n-butyl, may be prepared, e.g., by the esterification of compounds of the formula 104, which are described in Scheme 1, by conventional methods.
  • Compounds of the formula 134a and 134b are halogenated at the methyl position to produce either 3-bromomethyl-thiophene-2-carboxylic esters of the formula 136a or 2-bromomethyl-thiophene-3-carboxylic esters of the formula 136b by conventional methods; for example, addition of a halogen source (such as N-bromosuccinimide) and a radical initiator (such as benzoyl peroxide or azobisisobutyronitrile) in appropriate solvent (such as carbon tetrachloride, benzene, or dichloromethane), and allowing to react (thermally or UV initiation) for an appropriate length of time, where the thermal reaction of N-bromosuccinimide and benzoyl peroxide in carbon tetrachloride is preferred.
  • Alternative methods of this transformation may include, but are not limited to, oxidation of the methyl substituent to hydroxymethyl followed by conversion of the hydroxy group into an appropriate leaving group, such
  • Glycine derived reactants may include, but are not limited to, compounds with tert-butoxy carbonyl, tosyl, benzyl, or substituted benzyl groups (P) on the glycine nitrogen; and methyl, ethyl, or n-butyl groups (R) on the carboxylate; where N-(2,4-dimethoxy-benzyl)glycine ethyl ester is a preferred reactant.
  • Compounds of the formula 138a and 138b are converted to compounds of the formula 140a and 140b, respectively, by a series of steps in which intermediates may be isolated or may be used without purification.
  • compounds of the formula 138a and 138b are treated with an appropriate base, such as potassium tert-butoxide, lithium diisopropylamide, or lithium bis(trimethylsilyl)amide, to affect a Dieckman condensation.
  • the resulting compounds are then deprotected, using conventional conditions appropriate for the removal of the protecting group (P), and oxidized, using conventional conditions such as stirring under an oxygen-containing atmosphere or treating with an oxidizing agent such as bromine, to form compounds of the formula 140a and 140b.
  • the process of oxidation and deprotection may be done simultaneously, with use of a protecting group that is labile under the oxidative conditions.
  • One preferred method of this transformation is stirring a compound of formula 136a or 136b with N-(2,4-dimethoxy-benzyl)glycine ethyl ester in DMF with potassium carbonate to affect condensation, cyclizing with potassium tert-butoxide in THF, and then oxidizing and cleaving the N-2,4-dimethoxy-benzyl group by treatment with thionyl chloride in dichloromethane to provide compounds of the formula 140a or 140b, respectively.
  • Compounds of the formula 140a and 140b may be selectively halogenated to introduce a halogen alpha to the nitrogen of the pyridyl ring to produce compounds of the formula 122 and 124, respectively, where R 5 is halogen.
  • Conventional methods of halogenation may be used, such as treatment with N-bromosuccinimide or N-chlorosuccinamide in the presence of a radical initiator, or electrophilic reaction of N-bromosuccinimide in acetonitrile, where heating N-bromosuccinimide, benzoyl peroxide, and 140a or 140b in a carbon tetrachloride solution at reflux temperature for an appropriate period of time is preferred.
  • Compounds of the formula 140a and 140b may also be converted to compounds of formula 126 and 128 by conventional amidation conditions using a compound of the formula H 2 NCR 2 R 3 C(O)OH or an ester thereof, followed by appropriate de-esterification when necessary.
  • intermediates that contain one or more halogens may be modified by conventional means to transform the halogen into an alternate functional group.
  • examples may include, but are not limited to transformation of bromine into a hydrogen, cyano, alkoxy, aryloxy, thioether, acly, alkyl, alkenyl, aryl, or alkynyl substituent, which may occur at several points along the proceeding generic routes exemplified in schemes 1-3.
  • transformations may include, but are not limited to, Suzuki coupling reactions, Stille coupling reactions, Heck couplings, Castro-Stevens (Sonogashira) couplings, transition metal mediated carbonyl insertions, nucleophilic displacement, hydrogenation, metal exchange and electrophilic substitution.
  • these new functional groups may be chemically modified further through additional chemical transformations not shown on the general schemes.
  • transformation of halogens to an alternative substituent may occur on, but are not limited to, compounds of the formula 102, 104, 106, 108, 114, 116, 118, 120, 122, 124, 126, 128, 130, 132, 134a, 134b, 138a, 138b, 140a, and 140b, and compounds containing new substituents at the R 5 , R 6 , and/or R 7 positions may then be elaborated to final compounds following the generic synthetic schemes shown above. Sequential modification of compounds with multiple halogens may be employed to introduce non-equivalent substituents, such as exemplified in scheme 6.
  • Scheme 4 illustrates some specific conversion pathways to the synthesis of compounds of formula I, in particular how intermediates containing multiple halogens are converted to non-halogenated compounds.
  • Scheme 5 illustrates some specific conversion pathways to the synthesis of compounds of formula I, in particular how intermediates containing R 5 halogens are converted to non-halogenated compounds.
  • R 5 is a halogen, such as bromine
  • R 6 and R 7 are hydrogen
  • non-halogen species For example, treatment with appropriate nucleophilic species, such as copper cyanide, followed by conventional amidation methods are used to convert compounds 122 and 124 to compounds 160 and 162, respectively.
  • appropriate transition metal catalyst such as Pd(0) or Pd(II) reagents
  • organo-metal species for example organo tin reagents or organoborane reagents
  • Scheme 6 illustrates one example of a specific conversion pathway for the synthesis of compounds of formula I where intermediates containing halogens are converted to non-equivalent non-halogen substituents at R 5 , R 6 , and/or R 7 .
  • intermediates along schemes 1-3 that contain mono halogenation may be transformed by numerous conventional methods into non-halogen based intermediates.
  • Intermediates of type 122 and 124 may then undergo a second Suzuki coupling to introduce, e.g., a methyl group at the R 5 position.
  • Introduction of two independent halogen transforming reactions along general reaction schemes 1-3 allows for examples of formula I where, for example, R 6 and R 5 are not equal to halogen or hydrogen and are non-equivalent.
  • derivatization of the 3-hydroxyl group can proceed via conventional methods. For example, reaction of compounds 122, 124, 126, or 128 with a strong base such as lithium diisopropylamide (LDA) provides for an anionic oxygen substituent.
  • LDA lithium diisopropylamide
  • This intermediate is typically reacted in situ with a compound of the formula R′—Z (where R′ and the 3-oxygen of the pyridyl group provide for the R 1 group of formula I, and Z is a leaving group such as a halide) to provide for ether linkages.
  • Ether linkages at the 6-position of the pyridyl group can also proceed in the above manner provided that the stoichiometry is controlled to facilitate either mono etherification (at the 6-position) or di-etherification (at the 3- and 6-positions).
  • amination of the 6-halide proceeds under conventional conditions by contact with ammonia, a primary amine or a secondary amine.
  • oxidation of the sulfur atom in the thiophene ring to the corresponding sulfoxide or sulfone proceeds via conventional oxidation procedures including use of the appropriate stoichiometric amount of a suitable oxidizing agent such as m-chloroperbenzoic acid.
  • the compounds of the present invention can be used to modulate the stability and/or activity of HIF, and thereby activate HIF-regulated gene expression.
  • the compounds can be used in methods to treat, pretreat, or delay progression or onset of conditions associated with HIF including, but not limited to, anemic, ischemic, and hypoxic conditions.
  • the compound is administered immediately following a condition producing acute ischemia, e.g., myocardial infarction, pulmonary embolism, intestinal infarction, ischemic stroke, and renal ischemic-reperfusion injury.
  • the compound is administered to a patient diagnosed with a condition associated with the development of chronic ischemia, e.g., cardiac cirrhosis, macular degeneration, pulmonary embolism, acute respiratory failure, neonatal respiratory distress syndrome, and congestive heart failure.
  • the compound is administered immediately after a trauma or injury.
  • the compound can be administered to a subject based on predisposing conditions, e.g., hypertension, diabetes, occlusive arterial disease, chronic venous insufficiency, Raynaud's disease, chronic skin ulcers, cirrhosis, congestive heart failure, and systemic sclerosis.
  • compounds may be administered to pretreat a subject to decrease or prevent the development of tissue damage associated with ischemia or hypoxia.
  • the compounds of the present invention can be used to increase endogenous erythropoietin (EPO).
  • EPO endogenous erythropoietin
  • the compounds can be administered to prevent, pretreat, or treat EPO-associated conditions, including, e.g., conditions associated with anemia and neurological disorders.
  • Conditions associated with anemia include disorders such as acute or chronic kidney disease, diabetes, cancer, ulcers, infection with virus, e.g., UV, bacteria, or parasites; inflammation, etc.
  • Anemic conditions can further include those associated with procedures or treatments including, e.g., radiation therapy, chemotherapy, dialysis, and surgery.
  • Disorders associated with anemia additionally include abnormal hemoglobin and/or erythrocytes, such as found in disorders such as microcytic anemia, hypochromic anemia, aplastic anemia, etc.
  • the compounds can be used to increase endogenous EPO in a subject undergoing a specific treatment or procedure, prophylactically or concurrently, for example, an UV-infected anemic patient being treated with azidothymidine (zidovudine) or other reverse transcriptase inhibitors, an anemic cancer patient receiving cyclic cisplatin- or non-cisplatin-containing chemotherapeutics, or an anemic or non-anemic patient scheduled to undergo surgery. Additionally, the compounds can be used to increase endogenous EPO levels in an anemic or non-anemic patient scheduled to undergo surgery to reduce the need for allogenic blood transfusions or to facilitate banking of blood prior to surgery.
  • azidothymidine azidothymidine
  • the biological activity of the compounds of the invention may be assessed using any conventionally known methods. Suitable assay methods are well known in the art. The following assays are presented only as examples and are not intended to be limiting. The compounds of the invention are active in at least one of the following assays.
  • Human cells derived from various tissues are separately seeded into 35 mm culture dishes, and grown at 37° C., 20% O 2 , 5% CO 2 in standard culture medium, e.g., DMEM (Dulbecco's modification of Eagle's medium), 10% FBS (fetal bovine serum).
  • standard culture medium e.g., DMEM (Dulbecco's modification of Eagle's medium), 10% FBS (fetal bovine serum).
  • DMEM Dulbecco's modification of Eagle's medium
  • FBS fetal bovine serum
  • VEGF and EPO assays below.
  • the cells are washed two times in cold phosphate buffered saline (PBS) and then lysed in 1 ml of 10 mM Tris (pH 7.4), 1 mM EDTA, 150 mM NaCl, 0.5% IGEPAL (Sigma-Aldrich, St. Louis Mo.), and a protease inhibitor mix (Roche Molecular Biochemicals) for 15 minutes on ice.
  • Cell lysates are centrifuged at 3,000 ⁇ g for 5 minutes at 4° C., and the cytosolic fractions (supernatant) are collected.
  • the nuclei are resuspended and lysed in 100 ⁇ l of 20 mM HEPES (pH 7.2), 400 mM NaCl, 1 mM EDTA, 1 mM dithiothreitol, and a protease mix (Roche Molecular Biochemicals), centrifuged at 13,000 ⁇ g for 5 minutes at 4° C., and the nuclear protein fractions (supernatant) are collected.
  • Nuclear fractions are analyzed for HIF-1 ⁇ using a QUANTIKINE immunoassay (R&D Systems, Inc., Minneapolis Minn.) according to the manufacturer's instructions.
  • VEGF vascular endothelial growth factor
  • EPO erythropoietin
  • Ketoglutaric acid ⁇ -[1- 14 C]-sodium salt Ketoglutaric acid-[1- 14 C]-sodium salt, alpha-ketoglutaric acid sodium salt, and HPLC purified peptide may be obtained from commercial sources, e.g., Perkin-Elmer (Wellesley Mass.), Sigma-Aldrich, and SynPep Corp. (Dublin Calif.), respectively.
  • Peptides for use in the assay may be fragments of HIF ⁇ as described above or as disclosed in International Publication WO 2005/118836, incorporated by reference herein.
  • HIF-PH e.g., HIF-PH2 (EGLN1)
  • HIF-PH2 EGLN1
  • HIF-PH2 EGLN1
  • HIF-PH2 EGLN1
  • Enzyme activity is determined by capturing 14 CO 2 using an assay described by Kivirikko and Myllyla (1982, Methods Enzymol 82:245-304).
  • Assay reactions contain 50 mM HEPES (pH 7.4), 100 ⁇ M ⁇ -ketoglutaric acid sodium salt, 0.30 ⁇ Ci/ml ketoglutaric acid ⁇ -[1- 14 C]-sodium salt, 40 ⁇ M FeSO 4 , 1 mM ascorbate, 1541.8 units/ml Catalase, with or without 50 ⁇ M peptide substrate and various concentrations of compound of the invention. Reactions are initiated by addition of HIF-PH enzyme.
  • the peptide-dependent percent turnover is calculated by subtracting percent turnover in the absence of peptide from percent turnover in the presence of substrate peptide. Percent inhibition and IC 50 are calculated using peptide-dependent percent turnover at given inhibitor concentrations. Calculation of IC 50 values for each inhibitor is conducted using GraFit software (Erithacus Software Ltd., Surrey UK).
  • compositions of the present invention can be delivered directly or in pharmaceutical compositions along with suitable carriers or excipients, as is well known in the art.
  • Present methods of treatment can comprise administration of an effective amount of a compound of the invention to a subject having or at risk for anemia due to, e.g., chronic renal failure, diabetes, cancer, AIDS, radiation therapy, chemotherapy, kidney dialysis, or surgery.
  • the subject is a mammalian subject, and in a most preferred embodiment, the subject is a human subject.
  • Suitable routes of administration may, for example, include oral, rectal, topical, nasal, pulmonary, ocular, intestinal, and parenteral administration.
  • Primary routes for parenteral administration include intravenous, intramuscular, and subcutaneous administration.
  • Secondary routes of administration include intraperitoneal, intra-arterial, intra-articular, intracardiac, intracisternal, intradermal, intralesional, intraocular, intrapleural, intrathecal, intrauterine, and intraventricular administration.
  • the indication to be treated, along with the physical, chemical, and biological properties of the drug, dictate the type of formulation and the route of administration to be used, as well as whether local or systemic delivery would be preferred.
  • Pharmaceutical dosage forms of a compound of the invention may be provided in an instant release, controlled release, sustained release, or target drug-delivery system.
  • Commonly used dosage forms include, for example, solutions and suspensions, (micro-) emulsions, ointments, gels and patches, liposomes, tablets, dragees, soft or hard shell capsules, suppositories, ovules, implants, amorphous or crystalline powders, aerosols, and lyophilized formulations.
  • special devices may be required for application or administration of the drug, such as, for example, syringes and needles, inhalers, pumps, injection pens, applicators, or special flasks.
  • Pharmaceutical dosage forms are often composed of the drug, an excipient(s), and a container/closure system.
  • One or multiple excipients also referred to as inactive ingredients, can be added to a compound of the invention to improve or facilitate manufacturing, stability, administration, and safety of the drug, and can provide a means to achieve a desired drug release profile. Therefore, the type of excipient(s) to be added to the drug can depend on various factors, such as, for example, the physical and chemical properties of the drug, the route of administration, and the manufacturing procedure.
  • Pharmaceutically acceptable excipients are available in the art and include those listed in various pharmacopoeias. (See, e.g., the U.S.
  • compositions of the present invention can include one or more physiologically acceptable inactive ingredients that facilitate processing of active molecules into preparations for pharmaceutical use.
  • the composition may be formulated in aqueous solution, if necessary using physiologically compatible buffers, including, for example, phosphate, histidine, or citrate for adjustment of the formulation pH, and a tonicity agent, such as, for example, sodium chloride or dextrose.
  • physiologically compatible buffers including, for example, phosphate, histidine, or citrate for adjustment of the formulation pH
  • a tonicity agent such as, for example, sodium chloride or dextrose.
  • semisolid, liquid formulations, or patches may be preferred, possibly containing penetration enhancers.
  • penetration enhancers are generally known in the art.
  • the compounds can be formulated in liquid or solid dosage forms, and as instant or controlled/sustained release formulations.
  • Suitable dosage forms for oral ingestion by a subject include tablets, pills, dragees, hard and soft shell capsules, liquids, gels, syrups, slurries, suspensions, and emulsions.
  • the compounds may also be formulated in rectal compositions, such as suppositories or retention enemas, e.g., containing conventional suppository bases such as cocoa butter or other glycerides.
  • Solid oral dosage forms can be obtained using excipients, which may include fillers, disintegrants, binders (dry and wet), dissolution retardants, lubricants, glidants, antiadherants, cationic exchange resins, wetting agents, antioxidants, preservatives, coloring, and flavoring agents.
  • excipients may include fillers, disintegrants, binders (dry and wet), dissolution retardants, lubricants, glidants, antiadherants, cationic exchange resins, wetting agents, antioxidants, preservatives, coloring, and flavoring agents.
  • excipients can be of synthetic or natural source.
  • excipients examples include cellulose derivatives, citric acid, dicalcium phosphate, gelatine, magnesium carbonate, magnesium/sodium lauryl sulfate, mannitol, polyethylene glycol, polyvinyl pyrrolidone, silicates, silicium dioxide, sodium benzoate, sorbitol, starches, stearic acid or a salt thereof, sugars (i.e. dextrose, sucrose, lactose, etc.), talc, tragacanth mucilage, vegetable oils (hydrogenated), and waxes. Ethanol and water may serve as granulation aides.
  • coating of tablets with, for example, a taste-masking film, a stomach acid resistant film, or a release-retarding film is desirable.
  • Natural and synthetic polymers, in combination with colorants, sugars, and organic solvents or water, are often used to coat tablets, resulting in dragees.
  • the drug powder, suspension, or solution thereof can be delivered in a compatible hard or soft shell capsule.
  • the compounds of the present invention can be administered topically, such as through a skin patch, a semi-solid, or a liquid formulation, for example a gel, a (micro-) emulsion, an ointment, a solution, a (nano/micro)-suspension, or a foam.
  • a skin patch such as through a skin patch, a semi-solid, or a liquid formulation, for example a gel, a (micro-) emulsion, an ointment, a solution, a (nano/micro)-suspension, or a foam.
  • the penetration of the drug into the skin and underlying tissues can be regulated, for example, using penetration enhancers; the appropriate choice and combination of lipophilic, hydrophilic, and amphiphilic excipients, including water, organic solvents, waxes, oils, synthetic and natural polymers, surfactants, emulsifiers; by pH adjustment; and use of complexing agents.
  • Other techniques such as
  • the compounds for use according to the present invention are conveniently delivered in the form of a solution, suspension, emulsion, or semisolid aerosol from pressurized packs, or a nebuliser, usually with the use of a propellant, e.g., halogenated carbons derived from methane and Ethan, carbon dioxide, or any other suitable gas.
  • a propellant e.g., halogenated carbons derived from methane and Ethan, carbon dioxide, or any other suitable gas.
  • hydrocarbons like butane, isobutene, and pentane are useful.
  • the appropriate dosage unit may be determined by providing a valve to deliver a metered amount.
  • Capsules and cartridges of, for example, gelatin, for use in an inhaler or insufflator may be formulated. These typically contain a powder mix of the compound and a suitable powder base such as lactose or starch.
  • compositions formulated for parenteral administration by injection are usually sterile and, can be presented in unit dosage forms, e.g., in ampoules, syringes, injection pens, or in multi-dose containers, the latter usually containing a preservative.
  • the compositions may take such forms as suspensions, solutions, or emulsions in oily or aqueous vehicles, and may contain formulatory agents, such as buffers, tonicity agents, viscosity enhancing agents, surfactants, suspending and dispersing agents, antioxidants, biocompatible polymers, chelating agents, and preservatives.
  • the vehicle may contain water, a synthetic or vegetable oil, and/or organic co-solvents.
  • the parenteral formulation would be reconstituted or diluted prior to administration.
  • Depot formulations providing controlled or sustained release of a compound of the invention, may include injectable suspensions of nano/micro particles or nano/micro or non-micronized crystals.
  • Polymers such as poly(lactic acid), poly(glycolic acid), or copolymers thereof, can serve as controlled/sustained release matrices, in addition to others well known in the art.
  • Other depot delivery systems may be presented in form of implants and pumps requiring incision.
  • Suitable carriers for intravenous injection for the molecules of the invention are well-known in the art and include water-based solutions containing a base, such as, for example, sodium hydroxide, to form an ionized compound, sucrose or sodium chloride as a tonicity agent, for example, the buffer contains phosphate or histidine.
  • a base such as, for example, sodium hydroxide
  • sucrose or sodium chloride as a tonicity agent
  • the buffer contains phosphate or histidine.
  • Co-solvents such as, for example, polyethylene glycols, may be added.
  • These water-based systems are effective at dissolving compounds of the invention and produce low toxicity upon systemic administration.
  • the proportions of the components of a solution system may be varied considerably, without destroying solubility and toxicity characteristics.
  • the identity of the components may be varied.
  • low-toxicity surfactants such as polysorbates or poloxamers
  • polyethylene glycol or other co-solvents polyethylene glycol or other co-solvents
  • biocompatible polymers such as polyvinyl pyrrolidone may be added, and other sugars and polyols may substitute for dextrose.
  • a therapeutically effective dose can be estimated initially using a variety of techniques well-known in the art. Initial doses used in animal studies may be based on effective concentrations established in cell culture assays. Dosage ranges appropriate for human subjects can be determined, for example, using data obtained from animal studies and cell culture assays.
  • an effective amount or a therapeutically effective amount or dose of an agent refers to that amount of the agent or compound that results in amelioration of symptoms or a prolongation of survival in a subject.
  • Toxicity and therapeutic efficacy of such molecules can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., by determining the LD50 (the dose lethal to 50% of the population) and the ED50 (the dose therapeutically effective in 50% of the population).
  • the dose ratio of toxic to therapeutic effects is the therapeutic index, which can be expressed as the ratio LD50/ED50. Agents that exhibit high therapeutic indices are preferred.
  • the effective amount or therapeutically effective amount is the amount of the compound or pharmaceutical composition that will elicit the biological or medical response of a tissue, system, animal or human that is being sought by the researcher, veterinarian, medical doctor or other clinician. Dosages preferably fall within a range of circulating concentrations that includes the ED50 with little or no toxicity. Dosages may vary within this range depending upon the dosage form employed and/or the route of administration utilized. The exact formulation, route of administration, dosage, and dosage interval should be chosen according to methods known in the art, in view of the specifics of a subject's condition.
  • Dosage amount and interval may be adjusted individually to provide plasma levels of the active moiety that are sufficient to achieve the desired effects; i.e., the minimal effective concentration (MEC).
  • MEC minimal effective concentration
  • the MEC will vary for each compound but can be estimated from, for example, in vitro data and animal experiments. Dosages necessary to achieve the MEC will depend on individual characteristics and route of administration. In cases of local administration or selective uptake, the effective local concentration of the drug may not be related to plasma concentration.
  • the amount of agent or composition administered may be dependent on a variety of factors, including the sex, age, and weight of the subject being treated, the severity of the affliction, the manner of administration, and the judgment of the prescribing physician.
  • compositions may, if desired, be presented in a pack or dispenser device containing one or more unit dosage forms containing the active ingredient.
  • a pack or device may, for example, comprise metal or plastic foil, such as a blister pack, or glass, and rubber stoppers such as in vials.
  • the pack or dispenser device may be accompanied by instructions for administration.
  • Compositions comprising a compound of the invention formulated in a compatible pharmaceutical carrier may also be prepared, placed in an appropriate container, and labeled for treatment of an indicated condition.
  • the reaction was quenched with the addition of 10 g of sodium sulfite, and stirred for 15 min.
  • the mixture was acidified to ca. pH 3 with 1 N HCl solution, and cooled to 0° C.
  • An off-white precipitate was collected on a medium glass filter funnel.
  • the precipitate was washed twice with water and then dissolved in ethyl acetate.
  • the aqueous acetonitrile solution was extracted once with ethyl acetate, and the organic fraction was combined with the pervious ethyl acetate solution.
  • the combined organic fractions were dried over anhydrous magnesium sulfate and concentrated under hi-vacuum to produce 305 g of a tan solid.
  • reaction mixture was diluted with 10 mL ethyl acetate and 10 mL of saturated sodium bicarbonate solution was added.
  • the biphasic mixture was stirred for 15 min., filtered to remove insoluble materials, and separated to isolate the organic fraction.
  • the organic fraction was washed with brine, dried over anhydrous sodium sulfate, and concentrated under high vacuum to yield 74 mg of the mixture of title compounds.
  • the reaction mixture was cooled, filtered through a celite pad which was then washed twice with dicloromethane, and the combined organic fractions were concentrated to a residue under high vacuum.
  • the residue was dissolved in ethyl acetate and washed with a saturated sodium metabisulfite solution and a brine solution.
  • the organic fraction was dried over anhydrous sodium sulfate and concentrated to a solid residue under high vacuum.
  • the crude products were purified by flash chromatography eluting the two desired products with a gradient of 10-40% ethyl acetate in hexanes.
  • Product A first fraction, 7 mg; MS: (+) m/z 329.91, 331.90 (M+1, 79 Br/81Br);
  • Product B second fraction, 8 mg, MS: (+) m/z 329.92, 331.91 (M+1, 79 Br/ 81 Br)
  • the title compound was prepared from a mixture of 4,7-dihydroxy-2-(4-phenoxy-phenyl)-thieno[2,3-c]pyridine-5-carboxylic acid butyl ester and 4,7-dihydroxy-2-(4-phenoxy-phenyl)-thieno[3,2-c]pyridine-6-carboxylic acid butyl ester, example 7-b, under conditions analogous to experimental example 5-a.
  • the title compounds were further purified by column chromatography, eluting as a mixture from silica gel with a gradient of 10-30% ethyl acetate in hexanes. 1 H NMR of ca.
  • the reaction mixture was heated at 70° C. for 40 hours, cooled to room temperature, and diluted with ethyl acetate.
  • the organic mixture was successively washed with water, saturated sodium bicarbonate, and brine solutions.
  • the organic fractions were dried over anhydrous sodium sulfate and concentrated to a crude oily residue, which was then purified by column chromatography, eluting two major products from silica gel with a gradient of 10-50% ethyl acetate in hexanes: A (Higher R f product), 7 mg; MS: ( ⁇ ) m/z 432.30 (M ⁇ 1); B (Lower R f product), 11 mg; MS: ( ⁇ ) m/z 432.35 (M ⁇ 1).
  • the title compound was prepared from a mixture of 4-bromo-7-hydroxy-2-(4-phenoxy-phenyl)-thieno[3,2-c]pyridine-6-carboxylic acid butyl ester and 7-bromo-4-hydroxy-2-(4-phenoxy-phenyl)-thieno[2,3-c]pyridine-5-carboxylic acid butyl ester, example 7-c, under conditions analogous to experimental example 13-a with isolation of the higher R f isomer.
  • 5-Bromo-thiophene-2,3-dicarboxylic acid dimethyl ester (124 g, 444 mmol) was dissolved in 888 mL of methanol and cooled to 0° C. with an external ice bath. A solution of 222 mL of 2 N NaOH was added dropwise to the cold solution and stirred at 0° C. for 2 h. The reaction mixture was concentrated to ca. 300 mL and partitioned between 0.5 N HCl and ethyl acetate and extracted with ethyl acetate twice. The organic fractions were washed once with acidic brine solution, dried over anhydrous magnesium sulfate, and concentrated to a solid residue.
  • a portion of the crude intermediate (10 g, 28.6 mmol) was added to 120 mL of 0.5 N sodium n-butoxide in n-butanol.
  • the reaction mixture was heated at reflux temperature for 45 min, cooled, and 70 mL of 1 N HCl, 50 ml water and 20 mL ethyl acetate was added to precipitate the desired products.
  • the title compound was prepared from a mixture of 4,7-dihydroxy-2-phenylsulfanyl-thieno[3,2-c]pyridine-6-carboxylic acid n-butyl ester and 4,7 dihydroxy-2-phenylsulfanyl-thieno[2,3-c]pyridine-5-carboxylic acid n-butyl ester, example 17.a, under conditions analogous to experimental conditions 5.a.
  • the products were obtained as a mixture of the two isomers; MS: (+) m/z 437.91, 439.91 (M+1, 79 Br/ 81 Br).
  • the title compound was prepared from a mixture of 4-bromo-7-hydroxy-2-phenylsulfanyl-thieno[3,2-c]pyridine-6-carboxylic acid n-butyl ester and 7-bromo-4-hydroxy-2-phenylsulfanyl-thieno[2,3-c]pyridine-5-carboxylic acid n-butyl ester, example 17.b, under conditions analogous to experimental example 1.g.
  • the products were purified by column chromatography eluting from silica gel with a gradient of 20-60% ethyl acetate in hexanes.
  • Product A (Lower R f product); MS: (+) m/z 360.01 (M+1);
  • Product B (Higher R f product); MS: (+) m/z 360.02 (M+1).
  • 5-Bromo-3-methyl-thiophene-2-carboxylic acid (example 1.b, 10.0 g, 45.2 mmol) was suspended in 40 mL of thionyl chloride. The mixture was heated at reflux temperature for 1 hour and then cooled to 0° C. with an external ice bath. Cold ethyl alcohol (150 mL) was carefully added, and the solution was heated at reflux temperature for 16 hours. Under reduced pressure, the solution was concentrated to ca. one third of the original volume and then diluted with ethyl acetate and washed successively twice with saturated sodium bicarbonate solution, and once with brine.
  • 5-Bromo-3-methyl-thiophene-2-carboxylic acid ethyl ester (example 21.a, 5.0 g, 20.0 mmol), N-bromosuccinimide (3.65 g, 20.5 mmol), and benzoyl peroxide (484 mg, 2.00 mmol) were suspended in 50 mL of benzene and heated at reflux temperature for 16 hours. The reaction mixture was cooled and purified by column chromatography to elute 6.34 g of a crude mixture, which contained di-brominated product, from silica gel with a gradient of 0-20% ethyl acetate in hexanes.
  • ester 2-Bromo-7-hydroxy-thieno[3,2-c]pyridine-6-carboxylic acid ethyl ester 250 mg, 0.83 mmol, example 21.d
  • styreneboronic acid 185 mg, 1.25 mmol
  • cesium carbonate 675 mg, 2.08 mmol
  • tetrakis(triphenylphosphine)palladium(0) 92 mg, 0.08 mmol
  • the resultant mixture was diluted with dichloromethane and 2 N NaOH(aq) and filtered through a small pad of celite, the solid was washed with additional dichloromethane and NaOH solution.
  • the biphasic mixture was separated and the organic fraction was successively washed with 2 N NaOH and brine.
  • the organic fraction was dried over anhydrous magnesium sulfate and concentrated to a residue.
  • the crude product was purified by column chromatography, eluting the desired product from silica gel with a gradient of 5-30% ethyl acetate in hexanes to provide 6.4 g of product; MS: (+) m/z 219.4 (M+1)
  • the title compound was prepared from 3-methyl-5-phenoxy-thiophene-2-carboxylic acid, example 22.b, analogously to experimental example 21.a.
  • the product was isolated by column chromatography, eluting the desired product from silica gel with a gradient of 0-30% ethyl acetate in hexanes: MS: (+) m/z 262.9 (M+1)
  • the title compound was prepared from 2-cyano-7-hydroxy-thieno[3,2-c]pyridine-6-carboxylic acid ethyl ester, example 27.a, under conditions analogous to experimental example 17.d.
  • the crude precipitated product was further purified by dissolution in ethyl acetate and washing with water. The organic solution was dried over anhydrous sodium sulfate and concentrated under high vacuum to give the desired product; MS: ( ⁇ ) m/z 276.0 (M ⁇ 1)
  • N,N-Dimethylformamide 13 ⁇ L was added to a solution of oxalyl chloride (0.696 mL, 7.97 mmol), 4-bromo-thiophene-2,3-dicarboxylic acid 3-methyl ester and 4-bromo-thiophene-2,3-dicarboxylic acid 2-methyl ester (1.41 g, 5.32 mmol), example 30-c, in tetrahydrofuran (6.4 mL) at 0° C. The reaction mixture was stirred at 0° C. for 15 min, and then at ambient temperature for 1 h.
  • the crude product was purified by flash chromatography eluting from silica gel with a gradient of 0-60% ethyl acetate in hexanes to produce 1.0 g of 4-bromo-2-(ethoxycarbonylmethyl-carbamoyl)-thiophene-3-carboxylic acid methyl ester and 0.1 g of 4-bromo-3-(ethoxycarbonylmethyl-carbamoyl)thiophene-2-carboxylic acid methyl ester.
  • 4-bromo-2-(ethoxycarbonylmethyl-carbamoyl)-thiophene-3-carboxylic acid methyl ester, example 30-d (1.0 g, 2.86 mmol), 4-fluoro-phenyl boronic acid (0.479 mg, 3.43 mmol), potassium carbonate (1.42 g, 10.3 mmol), and tetrakis(triphenylphosphine)palladium(0) (495 mg, 0.43 mmol) were suspended in 45 mL of 1,4-dioxane. The reaction mixture was heated at reflux temperature overnight, cooled to room temperature and diluted with ethyl acetate.
  • the crude product was purified by column chromatography eluting from silica gel with a gradient of 0-50% ethyl acetate in dichloromethane to produce 78 mg of 3-(4-fluoro-phenyl)-4,7-dihydroxy-thieno[3,2-c]pyridine-6-carboxylic acid butyl ester and 252 mg of 3-(4-fluoro-phenyl)-4,7-dihydroxy-thieno[2,3-c]pyridine-5-carboxylic acid butyl ester.
US11/367,969 2005-03-02 2006-03-02 Thienopyridine compounds, and methods of use thereof Active 2031-04-22 US8703795B2 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US11/367,969 US8703795B2 (en) 2005-03-02 2006-03-02 Thienopyridine compounds, and methods of use thereof
US14/249,281 US9000006B2 (en) 2005-03-02 2014-04-09 Thienopyridine compounds, and methods of use thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US65813105P 2005-03-02 2005-03-02
US11/367,969 US8703795B2 (en) 2005-03-02 2006-03-02 Thienopyridine compounds, and methods of use thereof

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US14/249,281 Division US9000006B2 (en) 2005-03-02 2014-04-09 Thienopyridine compounds, and methods of use thereof

Publications (2)

Publication Number Publication Date
US20060199836A1 US20060199836A1 (en) 2006-09-07
US8703795B2 true US8703795B2 (en) 2014-04-22

Family

ID=36941909

Family Applications (2)

Application Number Title Priority Date Filing Date
US11/367,969 Active 2031-04-22 US8703795B2 (en) 2005-03-02 2006-03-02 Thienopyridine compounds, and methods of use thereof
US14/249,281 Active US9000006B2 (en) 2005-03-02 2014-04-09 Thienopyridine compounds, and methods of use thereof

Family Applications After (1)

Application Number Title Priority Date Filing Date
US14/249,281 Active US9000006B2 (en) 2005-03-02 2014-04-09 Thienopyridine compounds, and methods of use thereof

Country Status (6)

Country Link
US (2) US8703795B2 (zh)
EP (1) EP1866318B1 (zh)
JP (2) JP4792048B2 (zh)
CN (1) CN101166745A (zh)
AT (1) ATE513833T1 (zh)
WO (1) WO2006094292A2 (zh)

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8921389B2 (en) 2011-02-02 2014-12-30 Fibrogen, Inc. Naphthyridine derivatives as inhibitors of hypoxia inducible factor (HIF) hydroxylase
US9149476B2 (en) 2008-11-14 2015-10-06 Fibrogen, Inc. Thiochromene derivatives as HIF hydroxylase inhibitors
US9174976B2 (en) 2006-01-27 2015-11-03 Fibrogen, Inc. Cyanoisoquinoline compounds and methods of use thereof
US9340511B2 (en) 2012-07-16 2016-05-17 Fibrogen, Inc. Process for making isoquinoline compounds
US9371288B2 (en) 2012-07-16 2016-06-21 Fibrogen, Inc. Crystalline forms of a prolyl hydroxylase inhibitor
US9617218B2 (en) 2012-07-16 2017-04-11 Fibrogen, Inc. Crystalline forms of a prolyl hydroxylase inhibitor
US10065928B2 (en) 2014-09-02 2018-09-04 Sunshine Lake Pharma Co., Ltd. Quinolinone compound and use thereof
US10765672B2 (en) 2013-06-06 2020-09-08 Fibrogen, Inc. Pharmaceutical formulations of a HIF hydroxylase inhibitor
WO2021216530A1 (en) 2020-04-20 2021-10-28 Akebia Therapeutics, Inc. Treatment of viral infections, of organ injury, and of related conditions using a hif prolyl hydroxylase inhibitor or a hif-alpha stabilizer
WO2022150623A1 (en) 2021-01-08 2022-07-14 Akebia Therapeutics, Inc. Compounds and composition for the treatment of anemia
WO2022251563A1 (en) 2021-05-27 2022-12-01 Keryx Biopharmaceuticals, Inc. Pediatric formulations of ferric citrate

Families Citing this family (50)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7848703B1 (en) 2004-12-30 2010-12-07 Cypress Semiconductor Corporation Method and apparatus for binding wireless devices
US8614204B2 (en) 2003-06-06 2013-12-24 Fibrogen, Inc. Enhanced erythropoiesis and iron metabolism
EP2357175A1 (en) * 2003-06-06 2011-08-17 Fibrogen, Inc. isoquinoline derivatives AND THEIR USE IN INCREASING ENDOGENEOUS ERYTHROPOIETIN
WO2005019227A1 (en) * 2003-08-22 2005-03-03 Orchid Chemicals & Pharmaceuticals Ltd Process for the preparation of cephalosporin antibiotic
CA2610956A1 (en) * 2005-06-06 2006-12-14 Fibrogen, Inc. Improved treatment for anemia using a hif-alpha stabilising agent
GB0525164D0 (en) 2005-12-09 2006-01-18 Xention Discovery Ltd Compounds
KR20080097446A (ko) 2006-02-16 2008-11-05 피브로겐, 인크. 뇌졸중의 치료를 위한 화합물과 방법
CN101460497A (zh) * 2006-04-04 2009-06-17 菲布罗根有限公司 作为hif调节剂的吡咯并吡啶和噻唑并吡啶化合物
CN101186611B (zh) 2006-11-15 2011-05-18 天津和美生物技术有限公司 可抑制细胞释放肿瘤坏死因子的吡咯啉-2-酮衍生物及其制备和应用
CN101186612B (zh) * 2006-11-15 2012-10-03 天津和美生物技术有限公司 可抑制细胞释放肿瘤坏死因子的吡咯啉衍生物及其制备和应用
ES2393326T3 (es) 2006-12-18 2012-12-20 Amgen, Inc Compuestos basados en azaquinolona que presentan actividad inhibidora de prolil hidroxilasas, composiciones y usos de los mismos
EP2155680B1 (en) 2007-04-18 2013-12-04 Amgen, Inc Indanone derivatives that inhibit prolyl hydroxylase
US8048894B2 (en) 2007-04-18 2011-11-01 Amgen Inc. Quinolones and azaquinolones that inhibit prolyl hydroxylase
US8030346B2 (en) 2007-05-04 2011-10-04 Amgen Inc. Heterocyclic quinolone derivatives that inhibit prolyl hydroxylase activity
EP2155746A2 (en) 2007-05-04 2010-02-24 Amgen, Inc Diazaquinolones that inhibit prolyl hydroxylase activity
WO2009002533A1 (en) * 2007-06-27 2008-12-31 The Brigham And Women's Hospital, Inc. Inflammatory bowel disease therapies
TW200908984A (en) * 2007-08-07 2009-03-01 Piramal Life Sciences Ltd Pyridyl derivatives, their preparation and use
US20110028507A1 (en) * 2007-08-10 2011-02-03 Crystalgenomics, Inc. Pyridine derivatives and methods of use thereof
WO2009058403A1 (en) * 2007-11-02 2009-05-07 Fibrogen, Inc. Methods for reducing blood pressure
WO2009073669A1 (en) 2007-12-03 2009-06-11 Fibrogen, Inc. Isoxazolopyridine derivatives for use in the treatment of hif-mediated conditions
WO2009089547A1 (en) 2008-01-11 2009-07-16 Fibrogen, Inc. Isothiazole-pyridine derivatives as modulators of hif (hypoxia inducible factor) activity
WO2009100250A1 (en) 2008-02-05 2009-08-13 Fibrogen, Inc. Chromene derivatives and use thereof as hif hydroxylase activity inhibitors
CN101951775A (zh) * 2008-02-25 2011-01-19 默沙东公司 四氢噻吩并吡啶
CN102076680B (zh) 2008-04-28 2016-03-02 詹森药业有限公司 作为脯氨酰羟化酶抑制剂的苯并咪唑
CN102264740B (zh) 2008-08-20 2014-10-15 菲布罗根有限公司 一种吡咯[1,2-b]哒嗪衍生物及其作为hif调节剂的用途
WO2010024911A1 (en) * 2008-08-26 2010-03-04 Fibrogen, Inc. Methods for increasing neurogenesis
EP2412710A4 (en) 2009-03-27 2012-08-08 Kowa Co CONDENSED PIPERIDINE COMPOUND AND PHARMACEUTICAL AGENT CONTAINING THE SAME
GB201102659D0 (en) 2011-02-15 2011-03-30 Isis Innovation Assay
ITMI20110881A1 (it) * 2011-05-18 2012-11-19 E T C Srl Materiale semiconduttore organico
GB201113101D0 (en) 2011-07-28 2011-09-14 Isis Innovation Assay
TWI429652B (zh) 2011-08-05 2014-03-11 Ind Tech Res Inst 有機金屬化合物及包含其之有機電激發光裝置
WO2013043624A1 (en) 2011-09-23 2013-03-28 Merck Sharp & Dohme Corp. Substituted pyrimidines
EP2758058B1 (en) 2011-09-23 2017-04-26 Merck Sharp & Dohme Corp. Substituted pyrimidines
ES2689481T3 (es) 2011-10-25 2018-11-14 Janssen Pharmaceutica Nv Formulaciones de sal de meglumina de ácido 1-(5,6-dicloro-1h-benzo[d]imidazol-2-il)-1h-pirazol-4-carboxílico
US9446031B2 (en) 2012-01-18 2016-09-20 National University Of Singapore Compositions and methods for neovascularization
SG11201405574UA (en) 2012-03-09 2014-10-30 Fibrogen Inc 4 -hydroxy- isoquinoline compounds as hif hydroxylase inhibitors
ITMI20121691A1 (it) 2012-10-09 2014-04-10 E T C Srl Materiale semiconduttore organico
ITMI20121939A1 (it) * 2012-11-15 2014-05-16 E T C Srl Materiale organico semiconduttore
ITMI20121952A1 (it) 2012-11-16 2014-05-17 E T C Srl Materiale semiconduttore organico
CN104903295B (zh) * 2012-12-24 2017-09-01 卡迪拉保健有限公司 喹诺酮衍生物
CN105452227B (zh) 2013-01-24 2018-05-29 菲布罗根有限公司 {[1-氰基-5-(4-氯苯氧基)-4-羟基-异喹啉-3-羰基]-氨基}-乙酸的晶形
CN109180580B (zh) 2016-02-28 2021-01-12 深圳市塔吉瑞生物医药有限公司 一种取代的杂芳基化合物及包含该化合物的组合物及其用途
CA3031139A1 (en) 2016-07-19 2018-01-25 Bayer Cropscience Aktiengesellschaft Condensed bicyclic heterocycle derivatives as pest control agents
US20220054451A1 (en) * 2018-09-11 2022-02-24 Nikang Therapeutics, Inc. 2,3-DIHYDROBENZO[b]THIOPHENE DERIVATIVES AS HYPOXIA INDUCIBLE FACTOR-2(ALPHA) INHIBITORS
CN111499654B (zh) * 2019-01-30 2022-09-02 南通诺泰生物医药技术有限公司 一种合成3-氯-噻吩[2,3,-b]吡啶衍生物的方法
WO2020214853A1 (en) 2019-04-18 2020-10-22 Nikang Therapeutics, Inc. Tetrahydro-1h-cyclopenta[cd]indene derivatives as hypoxia inducible factor-2(alpha) inhibitors
CN110305143B (zh) * 2019-07-19 2021-03-09 济南新科医药科技有限公司 一种呋喃[2,3-c]并吡啶衍生物及其制备方法和用途
US11420936B2 (en) 2020-04-16 2022-08-23 Nikang Therapeutics, Inc. Hypoxia inducible factor-2(alpha) inhibitors and their use in the treatment of diseases
CN113234090B (zh) * 2021-04-23 2022-07-29 中国药科大学 靶向泛素化降解FoxM1的化合物或其可药用的盐、制备方法及用途
US20230101768A1 (en) * 2021-08-13 2023-03-30 The Board Of Regents Of The University Of Texas System Method to treat manganese toxicity and manganese-induced parkinsonism in humans

Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0650961A1 (de) 1993-11-02 1995-05-03 Hoechst Aktiengesellschaft Substituierte heterocyclische Carbonsäureamide, ihre Herstellung und ihre Verwendung als Arzneimittel
EP0661269A1 (de) 1993-12-30 1995-07-05 Hoechst Aktiengesellschaft Substituierte heterocyclische Carbonsäureamide, ihre Herstellung und ihre Verwendung als Arzneimittel
US5658933A (en) 1993-11-02 1997-08-19 Hoechst Aktiengesellschaft Substituted heterocyclic carboxamide esters, their preparation and their use as pharmaceuticals
WO2003010141A2 (en) 2001-07-25 2003-02-06 Boehringer Ingelheim (Canada) Ltd. Hepatitis c virus polymerase inhibitors with a heterobicyclic structure
WO2003014377A2 (en) 2001-08-07 2003-02-20 Boehringer Ingelheim (Canada) Ltd. Competitive binding assay for identifying inhibitors of hcv polymerase
WO2003053997A2 (en) 2001-12-06 2003-07-03 Fibrogen, Inc. Methods of increasing endogenous erythropoietin (epo)
WO2004108121A1 (en) 2003-06-06 2004-12-16 Fibrogen, Inc. Use of hif alpha stabilizers for enhancing erythropoiesis
WO2004108681A1 (en) 2003-06-06 2004-12-16 Fibrogen, Inc. Nitrogen-containing heteroaryl compounds and their use in increasing endogenous erythropoietin

Patent Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0650961A1 (de) 1993-11-02 1995-05-03 Hoechst Aktiengesellschaft Substituierte heterocyclische Carbonsäureamide, ihre Herstellung und ihre Verwendung als Arzneimittel
US5658933A (en) 1993-11-02 1997-08-19 Hoechst Aktiengesellschaft Substituted heterocyclic carboxamide esters, their preparation and their use as pharmaceuticals
EP0661269A1 (de) 1993-12-30 1995-07-05 Hoechst Aktiengesellschaft Substituierte heterocyclische Carbonsäureamide, ihre Herstellung und ihre Verwendung als Arzneimittel
WO2003010141A2 (en) 2001-07-25 2003-02-06 Boehringer Ingelheim (Canada) Ltd. Hepatitis c virus polymerase inhibitors with a heterobicyclic structure
WO2003014377A2 (en) 2001-08-07 2003-02-20 Boehringer Ingelheim (Canada) Ltd. Competitive binding assay for identifying inhibitors of hcv polymerase
WO2003053997A2 (en) 2001-12-06 2003-07-03 Fibrogen, Inc. Methods of increasing endogenous erythropoietin (epo)
WO2004108121A1 (en) 2003-06-06 2004-12-16 Fibrogen, Inc. Use of hif alpha stabilizers for enhancing erythropoiesis
WO2004108681A1 (en) 2003-06-06 2004-12-16 Fibrogen, Inc. Nitrogen-containing heteroaryl compounds and their use in increasing endogenous erythropoietin

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
Balant et al., "Metabolic Considerations, etc.," in Manfred ed. Burger's Medicinal Chemistry and Drug Discovery, 5rh ed. vol. 1: Principles and Practice, John Wiley & Sons, Inc., 1995. *
Ettmayer et al. "Lessons Learned from Marketed and Investigational Prodrugs." J. Med. Chem., (2004), 47(10): 2393-2404. *
Stella, Valentino. "Prodrugs as therapeutics." Expert Opin. Ther. Patents (2004), 14(3): 277-280. *
Testa, "Prodrug Research, etc.," Biochemical Pharmacology 68 (2004) 2097-2106. *

Cited By (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9174976B2 (en) 2006-01-27 2015-11-03 Fibrogen, Inc. Cyanoisoquinoline compounds and methods of use thereof
US9149476B2 (en) 2008-11-14 2015-10-06 Fibrogen, Inc. Thiochromene derivatives as HIF hydroxylase inhibitors
US9695170B2 (en) 2011-02-02 2017-07-04 Fibrogen, Inc. Naphthyridine derivatives as inhibitors of Hypoxia inducible factor (HIF) hydroxylase
US9271970B2 (en) 2011-02-02 2016-03-01 Fibrogen, Inc. Naphthyridine derivatives as inhibitors of hypoxia inducible factor (HIF) hydroxylase
US8921389B2 (en) 2011-02-02 2014-12-30 Fibrogen, Inc. Naphthyridine derivatives as inhibitors of hypoxia inducible factor (HIF) hydroxylase
US9708269B2 (en) 2012-07-16 2017-07-18 Fibrogen, Inc. Process for making isoquinoline compounds
US9617218B2 (en) 2012-07-16 2017-04-11 Fibrogen, Inc. Crystalline forms of a prolyl hydroxylase inhibitor
US9371288B2 (en) 2012-07-16 2016-06-21 Fibrogen, Inc. Crystalline forms of a prolyl hydroxylase inhibitor
US9340511B2 (en) 2012-07-16 2016-05-17 Fibrogen, Inc. Process for making isoquinoline compounds
US9918977B2 (en) 2012-07-16 2018-03-20 Fibrogen, Inc. Crystalline forms of a prolyl hydroxylase inhibitor
US10118897B2 (en) 2012-07-16 2018-11-06 Fibrogen, Inc. Crystalline forms of a prolyl hydroxylase inhibitor
US10272078B2 (en) 2012-07-16 2019-04-30 Fibrogen, Inc. Crystalline forms of a prolyl hydroxylase inhibitor
US10765672B2 (en) 2013-06-06 2020-09-08 Fibrogen, Inc. Pharmaceutical formulations of a HIF hydroxylase inhibitor
US10065928B2 (en) 2014-09-02 2018-09-04 Sunshine Lake Pharma Co., Ltd. Quinolinone compound and use thereof
WO2021216530A1 (en) 2020-04-20 2021-10-28 Akebia Therapeutics, Inc. Treatment of viral infections, of organ injury, and of related conditions using a hif prolyl hydroxylase inhibitor or a hif-alpha stabilizer
WO2022150623A1 (en) 2021-01-08 2022-07-14 Akebia Therapeutics, Inc. Compounds and composition for the treatment of anemia
WO2022251563A1 (en) 2021-05-27 2022-12-01 Keryx Biopharmaceuticals, Inc. Pediatric formulations of ferric citrate

Also Published As

Publication number Publication date
EP1866318A2 (en) 2007-12-19
US20060199836A1 (en) 2006-09-07
WO2006094292A3 (en) 2006-12-28
CN101166745A (zh) 2008-04-23
US9000006B2 (en) 2015-04-07
EP1866318B1 (en) 2011-06-22
ATE513833T1 (de) 2011-07-15
JP2011225583A (ja) 2011-11-10
WO2006094292A2 (en) 2006-09-08
JP4792048B2 (ja) 2011-10-12
US20140221422A1 (en) 2014-08-07
JP2008531736A (ja) 2008-08-14

Similar Documents

Publication Publication Date Title
US9000006B2 (en) Thienopyridine compounds, and methods of use thereof
US9387200B2 (en) Isothiazole-pyridine derivatives as modulators of HIF (hypoxia inducible factor) activity
US8269008B2 (en) Oxazolopyridine and isoxazolopyridine derivatives for use in the treatment of HIF-mediated conditions
US7696223B2 (en) Pyrrolo- and Thiazolo-pyridine compounds, and methods of use thereof
US9149476B2 (en) Thiochromene derivatives as HIF hydroxylase inhibitors
US8324405B2 (en) Chromene derivatives and use thereof as HIF hydroxylase activity inhibitors
US9695170B2 (en) Naphthyridine derivatives as inhibitors of Hypoxia inducible factor (HIF) hydroxylase
US8217043B2 (en) Compounds and methods for their use

Legal Events

Date Code Title Description
AS Assignment

Owner name: FIBROGEN, INC., CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:TURTLE, ERIC D.;FLIPPIN, LEE A.;AREND, MICHAEL P.;AND OTHERS;SIGNING DATES FROM 20061025 TO 20061116;REEL/FRAME:018566/0786

Owner name: FIBROGEN, INC., CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:TURTLE, ERIC D.;FLIPPIN, LEE A.;AREND, MICHAEL P.;AND OTHERS;REEL/FRAME:018566/0786;SIGNING DATES FROM 20061025 TO 20061116

STCF Information on status: patent grant

Free format text: PATENTED CASE

CC Certificate of correction
MAFP Maintenance fee payment

Free format text: PAYMENT OF MAINTENANCE FEE, 4TH YR, SMALL ENTITY (ORIGINAL EVENT CODE: M2551)

Year of fee payment: 4

FEPP Fee payment procedure

Free format text: ENTITY STATUS SET TO UNDISCOUNTED (ORIGINAL EVENT CODE: BIG.); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

MAFP Maintenance fee payment

Free format text: PAYMENT OF MAINTENANCE FEE, 8TH YEAR, LARGE ENTITY (ORIGINAL EVENT CODE: M1552); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

Year of fee payment: 8

AS Assignment

Owner name: WILMINGTON TRUST, NATIONAL ASSOCIATION, DELAWARE

Free format text: SECURITY INTEREST;ASSIGNOR:FIBROGEN, INC.;REEL/FRAME:063504/0221

Effective date: 20230429